Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Cell ; 36(4): 402-417.e13, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31564638

ABSTRACT

Metastasis is the leading cause of cancer mortality. Chromatin remodeling provides the foundation for the cellular reprogramming necessary to drive metastasis. However, little is known about the nature of this remodeling and its regulation. Here, we show that metastasis-inducing pathways regulate histone chaperones to reduce canonical histone incorporation into chromatin, triggering deposition of H3.3 variant at the promoters of poor-prognosis genes and metastasis-inducing transcription factors. This specific incorporation of H3.3 into chromatin is both necessary and sufficient for the induction of aggressive traits that allow for metastasis formation. Together, our data clearly show incorporation of histone variant H3.3 into chromatin as a major regulator of cell fate during tumorigenesis, and histone chaperones as valuable therapeutic targets for invasive carcinomas.


Subject(s)
Carcinoma/pathology , Chromatin/metabolism , Gene Expression Regulation, Neoplastic , Histones/metabolism , Neoplasm Metastasis/genetics , Animals , Carcinogenesis/genetics , Carcinoma/genetics , Cell Line, Tumor , Chromatin/genetics , Chromatin Assembly Factor-1/genetics , Chromatin Assembly Factor-1/metabolism , Disease Progression , Epigenesis, Genetic , Epithelial-Mesenchymal Transition/genetics , Female , Histones/genetics , Humans , Male , Mice , Promoter Regions, Genetic/genetics , RNA-Seq , Transcription Factors/genetics , Xenograft Model Antitumor Assays
2.
Cancer Res ; 77(18): 5077-5094, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28716898

ABSTRACT

Emerging observations link dysregulation of TANK-binding kinase 1 (TBK1) to developmental disorders, inflammatory disease, and cancer. Biochemical mechanisms accounting for direct participation of TBK1 in host defense signaling have been well described. However, the molecular underpinnings of the selective participation of TBK1 in a myriad of additional cell biological systems in normal and pathophysiologic contexts remain poorly understood. To elucidate the context-selective role of TBK1 in cancer cell survival, we employed a combination of broad-scale chemogenomic and interactome discovery strategies to generate data-driven mechanism-of-action hypotheses. This approach uncovered evidence that TBK1 supports AKT/mTORC1 pathway activation and function through direct modulation of multiple pathway components acting both upstream and downstream of the mTOR kinase itself. Furthermore, we identified distinct molecular features in which mesenchymal, Ras-mutant lung cancer is acutely dependent on TBK1-mediated support of AKT/mTORC1 pathway activation for survival. Cancer Res; 77(18); 5077-94. ©2017 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Mesoderm/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Small Molecule Libraries/pharmacology , TOR Serine-Threonine Kinases/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mesoderm/drug effects , Mesoderm/pathology , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Regulatory Elements, Transcriptional/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured
3.
Nat Chem Biol ; 11(6): 401-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25867045

ABSTRACT

Modern cancer treatment employs many effective chemotherapeutic agents originally discovered from natural sources. The cyclic depsipeptide didemnin B has demonstrated impressive anticancer activity in preclinical models. Clinical use has been approved but is limited by sparse patient responses combined with toxicity risk and an unclear mechanism of action. From a broad-scale effort to match antineoplastic natural products to their cellular activities, we found that didemnin B selectively induces rapid and wholesale apoptosis through dual inhibition of PPT1 and EEF1A1. Furthermore, empirical discovery of a small panel of exceptional responders to didemnin B allowed the generation of a regularized regression model to extract a sparse-feature genetic biomarker capable of predicting sensitivity to didemnin B. This may facilitate patient selection in a fashion that could enhance and expand the therapeutic application of didemnin B against neoplastic disease.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Depsipeptides/pharmacology , Membrane Proteins/antagonists & inhibitors , Peptide Elongation Factor 1/antagonists & inhibitors , Pharmacogenetics , Apoptosis/genetics , Biomarkers/metabolism , Cell Line, Tumor , Genome-Wide Association Study , Humans , Mechanistic Target of Rapamycin Complex 1 , Membrane Proteins/genetics , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Peptide Elongation Factor 1/genetics , Protein Biosynthesis/drug effects , Protein Biosynthesis/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Thiolester Hydrolases , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
4.
Cell ; 160(4): 715-728, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25679763

ABSTRACT

AMP-activated protein kinase (AMPK) is a master sensor and regulator of cellular energy status. Upon metabolic stress, AMPK suppresses anabolic and promotes catabolic processes to regain energy homeostasis. Cancer cells can occasionally suppress the growth-restrictive AMPK pathway by mutation of an upstream regulatory kinase. Here, we describe a widespread mechanism to suppress AMPK through its ubiquitination and degradation by the cancer-specific MAGE-A3/6-TRIM28 ubiquitin ligase. MAGE-A3 and MAGE-A6 are highly similar proteins normally expressed only in the male germline but frequently re-activated in human cancers. MAGE-A3/6 are necessary for cancer cell viability and are sufficient to drive tumorigenic properties of non-cancerous cells. Screening for targets of MAGE-A3/6-TRIM28 revealed that it ubiquitinates and degrades AMPKα1. This leads to inhibition of autophagy, activation of mTOR signaling, and hypersensitization to AMPK agonists, such as metformin. These findings elucidate a germline mechanism commonly hijacked in cancer to suppress AMPK.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antigens, Neoplasm/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Energy Metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Sequence Alignment , Signal Transduction , Testis/metabolism
5.
Sci Signal ; 6(297): ra90, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24129700

ABSTRACT

A challenge for biomedical research is the development of pharmaceuticals that appropriately target disease mechanisms. Natural products can be a rich source of bioactive chemicals for medicinal applications but can act through unknown mechanisms and can be difficult to produce or obtain. To address these challenges, we developed a new marine-derived, renewable natural products resource and a method for linking bioactive derivatives of this library to the proteins and biological processes that they target in cells. We used cell-based screening and computational analysis to match gene expression signatures produced by natural products to those produced by small interfering RNA (siRNA) and synthetic microRNA (miRNA) libraries. With this strategy, we matched proteins and miRNAs with diverse biological processes and also identified putative protein targets and mechanisms of action for several previously undescribed marine-derived natural products. We confirmed mechanistic relationships for selected siRNAs, miRNAs, and compounds with functional roles in autophagy, chemotaxis mediated by discoidin domain receptor 2, or activation of the kinase AKT. Thus, this approach may be an effective method for screening new drugs while simultaneously identifying their targets.


Subject(s)
Biological Products/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Ontology , Transcriptome/drug effects , Animals , Autophagy/drug effects , Autophagy/genetics , Bacteria/chemistry , Bacteria/classification , Biological Products/chemistry , Biological Products/isolation & purification , Cell Line, Tumor , Cells, Cultured , Cluster Analysis , Computational Biology/methods , Drug Evaluation, Preclinical/methods , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/genetics , HCT116 Cells , Humans , Invertebrates/chemistry , MCF-7 Cells , Marine Biology , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Molecular Structure , Oligonucleotide Array Sequence Analysis , RNA Interference
6.
Dev Cell ; 25(2): 182-95, 2013 Apr 29.
Article in English | MEDLINE | ID: mdl-23639443

ABSTRACT

Generating a balanced network of inhibitory and excitatory neurons during development requires precise transcriptional control. In the dorsal spinal cord, Ptf1a, a basic helix-loop-helix (bHLH) transcription activator, maintains this delicate balance by inducing homeodomain (HD) transcription factors such as Pax2 to specify the inhibitory lineage while suppressing HD factors such as Tlx1/3 that specify the excitatory lineage. We uncover the mechanism by which Ptf1a represses excitatory cell fate in the inhibitory lineage. We identify Prdm13 as a direct target of Ptf1a and reveal that Prdm13 actively represses excitatory cell fate by binding to regulatory sequences near the Tlx1 and Tlx3 genes to silence their expression. Prdm13 acts through multiple mechanisms, including interactions with the bHLH factor Ascl1, to repress Ascl1 activation of Tlx3. Thus, Prdm13 is a key component of a highly coordinated transcriptional network that determines the balance of inhibitory versus excitatory neurons in the dorsal spinal cord.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Neurons/metabolism , Spinal Cord/metabolism , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Differentiation , Cells, Cultured , Chick Embryo , Chromatin Immunoprecipitation , DNA Primers/chemistry , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fluorescent Antibody Technique , Homeodomain Proteins/genetics , In Situ Hybridization , Mice , Mice, Knockout , Neurons/cytology , PAX2 Transcription Factor/metabolism , RNA, Small Interfering/genetics , Spinal Cord/cytology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/physiology , Zinc Fingers
7.
J Exp Med ; 209(9): 1567-82, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22851595

ABSTRACT

Childhood herpes simplex virus-1 (HSV-1) encephalitis (HSE) may result from single-gene inborn errors of TLR3 immunity. TLR3-dependent induction of IFN-α/ß or IFN-λ is crucial for protective immunity against primary HSV-1 infection in the central nervous system (CNS). We describe here two unrelated children with HSE carrying different heterozygous mutations (D50A and G159A) in TBK1, the gene encoding TANK-binding kinase 1, a kinase at the crossroads of multiple IFN-inducing signaling pathways. Both mutant TBK1 alleles are loss-of-function but through different mechanisms: protein instability (D50A) or a loss of kinase activity (G159A). Both are also associated with an autosomal-dominant (AD) trait but by different mechanisms: haplotype insufficiency (D50A) or negative dominance (G159A). A defect in polyinosinic-polycytidylic acid-induced TLR3 responses can be detected in fibroblasts heterozygous for G159A but not for D50A TBK1. Nevertheless, viral replication and cell death rates caused by two TLR3-dependent viruses (HSV-1 and vesicular stomatitis virus) were high in fibroblasts from both patients, and particularly so in G159A TBK1 fibroblasts. These phenotypes were rescued equally well by IFN-α2b. Moreover, the IFN responses to the TLR3-independent agonists and viruses tested were maintained in both patients' peripheral blood mononuclear cells and fibroblasts. The narrow, partial cellular phenotype thus accounts for the clinical phenotype of these patients being limited to HSE. These data identify AD partial TBK1 deficiency as a new genetic etiology of childhood HSE, indicating that TBK1 is essential for the TLR3- and IFN-dependent control of HSV-1 in the CNS.


Subject(s)
Encephalitis, Herpes Simplex/genetics , Encephalitis, Herpes Simplex/immunology , Mutation , Protein Serine-Threonine Kinases/genetics , Toll-Like Receptor 3/immunology , Animals , Cell Death/immunology , Cells, Cultured , Child , Female , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/virology , Genes, Dominant , Herpesvirus 1, Human/pathogenicity , Humans , Interferon-beta/immunology , Male , Mice , Poly I-C/pharmacology , Protein Serine-Threonine Kinases/immunology , Vesiculovirus/pathogenicity
8.
Mol Cell ; 41(4): 458-70, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21329883

ABSTRACT

The innate immune-signaling kinase, TBK1, couples pathogen surveillance to induction of host defense mechanisms. Pathological activation of TBK1 in cancer can overcome programmed cell death cues, enabling cells to survive oncogenic stress. The mechanistic basis of TBK1 prosurvival signaling, however, has been enigmatic. Here, we show that TBK1 directly activates AKT by phosphorylation of the canonical activation loop and hydrophobic motif sites independently of PDK1 and mTORC2. Upon mitogen stimulation, triggering of the innate immune response, re-exposure to glucose, or oncogene activation, TBK1 is recruited to the exocyst, where it activates AKT. In cells lacking TBK1, insulin activates AKT normally, but AKT activation by exocyst-dependent mechanisms is impaired. Discovery and characterization of a 6-aminopyrazolopyrimidine derivative, as a selective low-nanomolar TBK1 inhibitor, indicates that this regulatory arm can be pharmacologically perturbed independently of canonical PI3K/PDK1 signaling. Thus, AKT is a direct TBK1 substrate that connects TBK1 to prosurvival signaling.


Subject(s)
Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Cell Survival , Cell Transformation, Neoplastic , Cells, Cultured , HCT116 Cells , Humans , Immunity, Innate , Mice , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Transfection
9.
Cell ; 144(2): 253-67, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21241894

ABSTRACT

The study of macroautophagy in mammalian cells has described induction, vesicle nucleation, and membrane elongation complexes as key signaling intermediates driving autophagosome biogenesis. How these components are recruited to nascent autophagosomes is poorly understood, and although much is known about signaling mechanisms that restrain autophagy, the nature of positive inductive signals that can promote autophagy remain cryptic. We find that the Ras-like small G protein, RalB, is localized to nascent autophagosomes and is activated on nutrient deprivation. RalB and its effector Exo84 are required for nutrient starvation-induced autophagocytosis, and RalB activation is sufficient to promote autophagosome formation. Through direct binding to Exo84, RalB induces the assembly of catalytically active ULK1 and Beclin1-VPS34 complexes on the exocyst, which are required for isolation membrane formation and maturation. Thus, RalB signaling is a primary adaptive response to nutrient limitation that directly engages autophagocytosis through mobilization of the core vesicle nucleation machinery.


Subject(s)
Autophagy , Epithelial Cells/pathology , Phagosomes/metabolism , Signal Transduction , ral GTP-Binding Proteins/metabolism , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cell Line , Class III Phosphatidylinositol 3-Kinases/metabolism , Epithelial Cells/microbiology , Humans , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Salmonella typhimurium/physiology , Stress, Physiological , Vesicular Transport Proteins/metabolism
10.
EMBO J ; 26(17): 3968-80, 2007 Sep 05.
Article in English | MEDLINE | ID: mdl-17690688

ABSTRACT

Proper regulation of cell cycle progression is pivotal for maintaining genome stability. In a search for DNA damage-inducible, CHK1-modulated genes, we have identified BTG3 (B-cell translocation gene 3) as a direct p53 target. The p53 transcription factor binds to a consensus sequence located in intron 2 of the gene both in vitro and in vivo, and depletion of p53 by small interfering RNA (siRNA) abolishes DNA damage-induced expression of the gene. Furthermore, ablation of BTG3 by siRNA in cancer cells results in accelerated exit from the DNA damage-induced G2/M block. In vitro, BTG3 binds to and inhibits E2F1 through an N-terminal domain including the conserved box A. Deletion of the interaction domain in BTG3 abrogates not only its growth suppression activity, but also its repression on E2F1-mediated transactivation. We also present evidence that by disrupting the DNA binding activity of E2F1, BTG3 participates in the regulation of E2F1 target gene expression. Therefore, our studies have revealed a previously unidentified pathway through which the activity of E2F1 may be guarded by activated p53.


Subject(s)
E2F1 Transcription Factor/metabolism , Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Cycle Proteins , Cell Line, Tumor , DNA Damage , E2F1 Transcription Factor/antagonists & inhibitors , Humans , Introns , Oligonucleotide Array Sequence Analysis , Protein Binding , Proteins/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
11.
Mol Biol Cell ; 16(4): 1684-95, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15659650

ABSTRACT

The tumor suppressor protein p53 mediates stress-induced growth arrest or apoptosis and plays a major role in safeguarding genome integrity. In response to DNA damage, p53 can be modified at multiple sites by phosphorylation and acetylation. We report on the characterization of p53 C-terminal phosphorylation by CHK1 and CHK2, two serine/threonine (Ser/Thr) protein kinases, previously implicated in the phosphorylation of the p53 N terminus. Using tryptic phosphopeptide mapping, we have identified six additional CHK1 and CHK2 sites residing in the final 100 amino acids of p53. Phosphorylation of at least three of these sites, Ser366, Ser378, and Thr387, was induced by DNA damage, and the induction at Ser366 and Thr387 was abrogated by small interfering RNA targeting chk1 and chk2. Furthermore, mutation of these phosphorylation sites has a different impact on p53 C-terminal acetylation and on the activation of p53-targeted promoters. Our results demonstrate a possible interplay between p53 C-terminal phosphorylation and acetylation, and they provide an additional mechanism for the control of the activity of p53 by CHK1 and CHK2.


Subject(s)
DNA Damage , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Acetylation , Cell Line, Tumor , Checkpoint Kinase 1 , Checkpoint Kinase 2 , Down-Regulation , Humans , Lysine/genetics , Lysine/metabolism , Mutation/genetics , Phosphorylation , Phosphoserine/metabolism , Phosphothreonine/metabolism , Promoter Regions, Genetic/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/genetics
12.
J Biol Chem ; 280(9): 7748-57, 2005 Mar 04.
Article in English | MEDLINE | ID: mdl-15618221

ABSTRACT

CHK2/hCds1 plays important roles in the DNA damage-induced cell cycle checkpoint by phosphorylating several important targets, such as Cdc25 and p53. To obtain a better understanding of the CHK2 signaling pathway, we have carried out a yeast two-hybrid screen to search for potential CHK2-interacting proteins. Here, we report the identification of the mitotic checkpoint kinase, TTK/hMps1, as a novel CHK2-interacting protein. TTK/hMps1 directly phosphorylates CHK2 on Thr-68 in vitro. Expression of a TTK kinase-dead mutant, TTK(D647A), interferes with the G(2)/M arrest induced by either ionizing radiation or UV light. Interestingly, induction of CHK2 Thr-68 phosphorylation and of several downstream events, such as cyclin B1 accumulation and Cdc2 Tyr-15 phosphorylation, is also affected. Furthermore, ablation of TTK expression using small interfering RNA results not only in reduced CHK2 Thr-68 phosphorylation, but also in impaired growth arrest. Our results are consistent with a model in which TTK functions upstream from CHK2 in response to DNA damage and suggest possible cross-talk between the spindle assembly checkpoint and the DNA damage checkpoint.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle Proteins/physiology , DNA Damage , Protein Kinases/metabolism , Protein Kinases/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/physiology , Threonine/chemistry , Blotting, Western , Cell Cycle , Cell Division , Cell Line , Cell Line, Tumor , Checkpoint Kinase 2 , Cyclin B/metabolism , Cyclin B1 , Escherichia coli/metabolism , G2 Phase , Glutathione Transferase/metabolism , HeLa Cells , Humans , Immunoprecipitation , Models, Biological , Mutation , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases , RNA, Small Interfering/metabolism , Radiation, Ionizing , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transfection , Tumor Suppressor Protein p53/metabolism , Two-Hybrid System Techniques , Tyrosine/chemistry , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...